Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Biophys J ; 117(9): 1751-1763, 2019 11 05.
Article in English | MEDLINE | ID: mdl-31587826

ABSTRACT

Obstructing conductive pathways of the channel-forming toxins with targeted blockers is a promising drug design approach. Nearly all tested positively charged ligands have been shown to reversibly block the cation-selective channel-forming protective antigen (PA63) component of the binary anthrax toxin. The cationic ligands with more hydrophobic surfaces, particularly those carrying aromatic moieties, inhibited PA63 more effectively. To understand the physical basis of PA63 selectivity for a particular ligand, detailed information is required on how the blocker structural elements (e.g., positively charged and aromatic groups) influence the molecular kinetics of the blocker/channel binding reactions. In this study, we address this problem using the high-resolution single-channel planar lipid bilayer technique. Several structurally distinct cationic blockers, namely per-6-S-(3-amino) propyl-ß-cyclodextrin, per-6-S-(3-aminomethyl) benzyl-α-cyclodextrin, per-6-S-(3-aminomethyl) benzyl-ß-cyclodextrin, per-6-S-(3-aminomethyl) benzyl-γ-cyclodextrin, methyltriphenylphosphonium ion, and G0 polyamidoamine dendrimer are tested for their ability to inhibit the heptameric and octameric PA63 variants and PA63F427A mutant. The F427 residues form a hydrophobic constriction region inside the channel, known as the "ϕ-clamp." We show that the cationic blockers interact with PA63 through a combination of forces. Analysis of the binding reaction kinetics suggests the involvement of cation-π, Coulomb, and salt-concentration-independent π-π or hydrophobic interactions in the cationic cyclodextrin binding. It is possible that these blockers bind to the ϕ-clamp and are also stabilized by the Coulomb interactions of their terminal amino groups with the water-exposed negatively charged channel residues. In PA63F427A, only the suggested Coulomb component of the cyclodextrin interaction remains. Methyltriphenylphosphonium ion and G0 polyamidoamine dendrimer, despite being positively charged, interact primarily with the ϕ-clamp. We also show that seven- and eightfold symmetric cyclodextrins effectively block the heptameric and octameric forms of PA63 interchangeably, adding flexibility to the earlier formulated blocker/target symmetry match requirement.


Subject(s)
Antigens, Bacterial/chemistry , Bacterial Toxins/chemistry , Cations , Dendrimers/chemistry , Kinetics , Onium Compounds/chemistry , Time Factors , Trityl Compounds/chemistry , beta-Cyclodextrins/chemistry
2.
Int J Pharm ; 531(2): 714-717, 2017 Oct 15.
Article in English | MEDLINE | ID: mdl-28750897

ABSTRACT

Clostridium perfringens epsilon toxin (ETX) is considered as one of the most dangerous potential biological weapons. The goal of this work was to identify inhibitors of ETX using a novel approach for the inactivation of pore-forming toxins. The approach is based on the blocking of the target pore with molecules having the same symmetry as the pore itself. About 200 various ß-cyclodextrin derivatives were screened for inhibitors of ETX activity using a colorimetric cell viability assay. Several compounds with dose-dependent activities at low micromolar concentrations have been identified. The same compounds were also able to inhibit lethal toxin of Bacillus anthracis.


Subject(s)
Bacterial Toxins/antagonists & inhibitors , Clostridium perfringens/drug effects , beta-Cyclodextrins/pharmacology , Bacillus anthracis/drug effects
3.
Toxins (Basel) ; 6(7): 2097-114, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-25029374

ABSTRACT

Some Clostridium difficile strains produce, in addition to toxins A and B, the binary toxin Clostridium difficile transferase (CDT), which ADP-ribosylates actin and may contribute to the hypervirulence of these strains. The separate binding and translocation component CDTb mediates transport of the enzyme component CDTa into mammalian target cells. CDTb binds to its receptor on the cell surface, CDTa assembles and CDTb/CDTa complexes are internalised. In acidic endosomes, CDTb mediates the delivery of CDTa into the cytosol, most likely by forming a translocation pore in endosomal membranes. We demonstrate that a seven-fold symmetrical positively charged ß-cyclodextrin derivative, per-6-S-(3-aminomethyl)benzylthio-ß-cyclodextrin, which was developed earlier as a potent inhibitor of the translocation pores of related binary toxins of Bacillus anthracis, Clostridium botulinum and Clostridium perfringens, protects cells from intoxication with CDT. The pore blocker did not interfere with the CDTa-catalyzed ADP-ribosylation of actin or toxin binding to Vero cells but inhibited the pH-dependent membrane translocation of CDTa into the cytosol. In conclusion, the cationic ß-cyclodextrin could serve as the lead compound in a development of novel pharmacological strategies against the CDT-producing strains of C. difficile.


Subject(s)
ADP Ribose Transferases/toxicity , Clostridioides difficile , Protective Agents/pharmacology , beta-Cyclodextrins/pharmacology , Animals , Cell Membrane/drug effects , Cell Membrane/metabolism , Chlorocebus aethiops , Vero Cells
4.
Int J Pharm ; 458(1): 25-30, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-24157345

ABSTRACT

This study reports the characterization of three cationic amphiphillic aminocalix[4]arenes as potential antimicrobial agents in vitro. In cytotoxicity tests on mouse macrophage RAW 264.7 cells aminocalix[4]arenes 1 and 3 showed no toxicity up to 200 and 100 µM concentrations, respectively, while 2 was non-toxic only up to 50 µM. With regard to the haemolytic activity on rabbit red blood cells, 1 was not active at concentrations up to 100 µM in contrast to the other two studied macrocycles. Compounds showed negligible ability to protect either mouse macrophage RAW 264.7 cells from anthrax lethal toxin of Bacillus anthracis (B. anthracis) or rabbit red blood cells from α-haemolysin of Staphylococcus aureus (S. aureus) in comparison to amino-ß-cyclodextrins. However, all aminocalix[4]arenes showed potential as antimicrobials. Their minimum inhibitory concentrations (MIC) against Escherichia coli (E. coli) and S. aureus were in the 16-32 µg/ml concentration range, while minimum lethal concentrations (MLC) varied from 16 to 256 µg/ml depending on the bacteria and aminocalix[4]arene considered. Macrocycle 1 showed partial synergism against S. aureus in tandem with a model antibacterial drug, fusidic acid, at certain concentration combinations.


Subject(s)
Anti-Bacterial Agents/pharmacology , Cations/pharmacology , Animals , Antigens, Bacterial , Bacillus anthracis/drug effects , Bacterial Toxins , Cell Line , Escherichia coli/drug effects , Macrophages/drug effects , Mice , Microbial Sensitivity Tests/methods , Rabbits , Staphylococcus aureus/drug effects , beta-Cyclodextrins/pharmacology
5.
Curr Opin Pharmacol ; 13(5): 717-25, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24011515

ABSTRACT

Cyclodextrin derivatives can be utilized as anti-infectives with pore-forming proteins as the targets. The highly efficient selection of potent inhibitors was achieved because per-substituted cyclodextrins have the same symmetry as the target pores. Inhibitors of several bacterial toxins produced by Bacillus anthracis, Staphylococcus aureus, Clostridium perfringens, Clostridium botulinum, and Clostridium difficile were identified from a library of ∼200 CD derivatives. It was demonstrated that multi-targeted inhibitors can be found using this approach and could be utilized for the development of broad-spectrum drugs against various pathogens.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Bacterial Toxins/antagonists & inhibitors , Cyclodextrins/therapeutic use , Animals , Anti-Bacterial Agents/pharmacology , Bacillus anthracis , Bacterial Toxins/metabolism , Clostridium perfringens , Cyclodextrins/pharmacology , Humans , Staphylococcus aureus
6.
Proc Natl Acad Sci U S A ; 109(45): 18453-8, 2012 Nov 06.
Article in English | MEDLINE | ID: mdl-23100532

ABSTRACT

The thermodynamics of binding reactions is usually studied in the framework of the linear van't Hoff analysis of the temperature dependence of the equilibrium constant. The logarithm of the equilibrium constant is plotted versus inverse temperature to discriminate between two terms: an enthalpic contribution that is linear in the inverse temperature, and a temperature-independent entropic contribution. When we apply this approach to a particular case-blockage of the anthrax PA(63) channel by a multicharged cyclodextrin derivative-we obtain a nearly linear behavior with a slope that is characterized by enthalpy of about 1 kcal/mol. In contrast, from blocker partitioning between the channel and the bulk, we estimate the depth of the potential well for the blocker in the channel to be at least 8 kcal/mol. To understand this apparent discrepancy, we use a simple model of particle interaction with the channel and show that this significant difference between the two estimates is due to the temperature dependence of the physical forces between the blocker and the channel. In particular, we demonstrate that if the major component of blocker-channel interaction is van der Waals interactions and/or Coulomb forces in water, the van't Hoff enthalpy of the binding reaction may be close to zero or even negative, including cases of relatively strong binding. The results are quite general and, therefore, of importance for studies of enzymatic reactions, rational drug design, small-molecule binding to proteins, protein-protein interactions, and protein folding, among others.


Subject(s)
Antigens, Bacterial/chemistry , Antigens, Bacterial/metabolism , Bacterial Toxins/chemistry , Bacterial Toxins/metabolism , Cyclodextrins/pharmacology , Ion Channels/antagonists & inhibitors , Cations , Ion Channel Gating/drug effects , Ion Channels/metabolism , Kinetics , Protein Binding/drug effects , Solutions , Thermodynamics
7.
Biophys J ; 103(6): 1208-17, 2012 Sep 19.
Article in English | MEDLINE | ID: mdl-22995493

ABSTRACT

Cationic ß-cyclodextrin derivatives were recently introduced as highly effective, potentially universal blockers of three binary bacterial toxins: anthrax toxin of Bacillus anthracis, C2 toxin of Clostridium botulinum, and iota toxin of Clostridium perfringens. The binary toxins are made of two separate components: the enzymatic A component, which acts on certain intracellular targets, and the binding/translocation B component, which forms oligomeric channels in the target cell membrane. Here we studied the voltage and salt dependence of the rate constants of binding and dissociation reactions of two structurally different ß-cyclodextrins (AmPrßCD and AMBnTßCD) in the PA(63), C2IIa, and Ib channels (B components of anthrax, C2, and iota toxins, respectively). With all three channels, the blocker carrying extra hydrophobic aromatic groups on the thio-alkyl linkers of positively charged amino groups, AMBnTßCD, demonstrated significantly stronger binding compared with AmPrßCD. This effect is seen as an increased residence time of the blocker in the channels, whereas the time between blockages characterizing the binding reaction on-rate stays practically unchanged. Surprisingly, the voltage sensitivity, expressed as a slope of the logarithm of the blocker residence time as a function of voltage, turned out to be practically the same for all six cases studied, suggesting structural similarities among the three channels. Also, the more-effective AMBnTßCD blocker shows weaker salt dependence of the binding and dissociation rate constants compared with AmPrßCD. By estimating the relative contributions of the applied transmembrane field, long-range Coulomb, and salt-concentration-independent, short-range forces, we found that the latter represent the leading interaction, which accounts for the high efficiency of blockage. In a search for the putative groups in the channel lumen that are responsible for the short-range forces, we performed measurements with the F427A mutant of PA(63), which lacks the functionally important phenylalanine clamp. We found that the on-rates of the blockage were virtually conserved, but the residence times and, correspondingly, the binding constants dropped by more than an order of magnitude, which also reduced the difference between the efficiencies of the two blockers.


Subject(s)
Bacillus anthracis/metabolism , Bacterial Toxins/antagonists & inhibitors , Bacterial Toxins/metabolism , Clostridium botulinum/metabolism , Clostridium perfringens/metabolism , beta-Cyclodextrins/metabolism , beta-Cyclodextrins/pharmacology , Bacillus anthracis/cytology , Bacterial Toxins/genetics , Biological Transport/drug effects , Clostridium botulinum/cytology , Clostridium perfringens/cytology , Dose-Response Relationship, Drug , Electric Conductivity , Kinetics , Mutation , Porosity , Potassium Chloride/pharmacology , Protein Binding , beta-Cyclodextrins/chemistry
8.
PLoS One ; 6(8): e23927, 2011.
Article in English | MEDLINE | ID: mdl-21887348

ABSTRACT

BACKGROUND: Clostridium botulinum C2 toxin and Clostridium perfringens iota toxin are binary exotoxins, which ADP-ribosylate actin in the cytosol of mammalian cells and thereby destroy the cytoskeleton. C2 and iota toxin consists of two individual proteins, an enzymatic active (A-) component and a separate receptor binding and translocation (B-) component. The latter forms a complex with the A-component on the surface of target cells and after receptor-mediated endocytosis, it mediates the translocation of the A-component from acidified endosomal vesicles into the cytosol. To this end, the B-components form heptameric pores in endosomal membranes, which serve as translocation channels for the A-components. METHODOLOGY/PRINCIPAL FINDINGS: Here we demonstrate that a 7-fold symmetrical positively charged ß-cyclodextrin derivative, per-6-S-(3-aminomethyl)benzylthio-ß-cyclodextrin, protects cultured cells from intoxication with C2 and iota toxins in a concentration-dependent manner starting at low micromolar concentrations. We discovered that the compound inhibited the pH-dependent membrane translocation of the A-components of both toxins in intact cells. Consistently, the compound strongly blocked transmembrane channels formed by the B-components of C2 and iota toxin in planar lipid bilayers in vitro. With C2 toxin, we consecutively ruled out all other possible inhibitory mechanisms showing that the compound did not interfere with the binding of the toxin to the cells or with the enzyme activity of the A-component. CONCLUSIONS/SIGNIFICANCE: The described ß-cyclodextrin derivative was previously identified as one of the most potent inhibitors of the binary lethal toxin of Bacillus anthracis both in vitro and in vivo, implying that it might represent a broad-spectrum inhibitor of binary pore-forming exotoxins from pathogenic bacteria.


Subject(s)
Clostridium botulinum/pathogenicity , Clostridium perfringens/pathogenicity , Exotoxins/antagonists & inhibitors , beta-Cyclodextrins/pharmacology , Biological Transport/drug effects , Cell Line , Cytosol/metabolism , Dose-Response Relationship, Drug , Endocytosis/drug effects , Endosomes/metabolism , Exotoxins/metabolism , Humans , Lipid Bilayers/metabolism , Protective Agents , beta-Cyclodextrins/therapeutic use
9.
Antimicrob Agents Chemother ; 55(7): 3594-7, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21555769

ABSTRACT

We compared the abilities of structurally related cationic cyclodextrins to inhibit Bacillus anthracis lethal toxin and Staphylococcus aureus α-hemolysin. We found that both ß- and γ-cyclodextrin derivatives effectively inhibited anthrax toxin action by blocking the transmembrane oligomeric pores formed by the protective antigen (PA) subunit of the toxin, whereas α-cyclodextrins were ineffective. In contrast, α-hemolysin was selectively blocked only by ß-cyclodextrin derivatives, demonstrating that both symmetry and size of the inhibitor and the pore are important.


Subject(s)
Bacterial Toxins/chemistry , alpha-Cyclodextrins/chemistry , beta-Cyclodextrins/chemistry , gamma-Cyclodextrins/chemistry , Animals , Antigens, Bacterial/chemistry , Cell Death/drug effects , Cell Line , Hemolysin Proteins/chemistry , Molecular Conformation , Staphylococcus aureus/metabolism
10.
ChemMedChem ; 6(1): 181-92, 2011 Jan 03.
Article in English | MEDLINE | ID: mdl-21140396

ABSTRACT

Three new series of potential anthrax toxin inhibitors based on the ß-cyclodextrin (ßCD) scaffold were developed by exploiting face-selective Cu(I)-catalyzed azide-alkyne 1,3-cycloadditions, amine-isothiocyanate coupling, and allyl group hydroboration-oxidation/hydroxy → amine replacement reactions. The molecular design follows the "symmetry-complementarity" concept between homogeneously functionalized polycationic ßCD derivatives and protective antigen (PA), a component of anthrax toxin known to form C7-symmetric pores on the cell membrane used by lethal and edema factors to gain access to the cytosol. The synthesis and antitoxin activity of a collection of ßCD derivatives differing in the number, arrangement, and face location of the cationic elements are reported herein. These results set the basis for a structure-activity relationship development program of new candidates to combat the anthrax threat.


Subject(s)
Antigens, Bacterial , Bacterial Toxins , Polyamines , beta-Cyclodextrins , Animals , Anthrax/drug therapy , Anthrax/immunology , Anthrax/metabolism , Antigens, Bacterial/immunology , Antigens, Bacterial/metabolism , Bacillus anthracis/immunology , Bacillus anthracis/metabolism , Bacterial Toxins/antagonists & inhibitors , Bacterial Toxins/immunology , Bacterial Toxins/metabolism , Cell Line , Chemistry, Pharmaceutical , Cluster Analysis , Computer-Aided Design , Mice , Models, Molecular , Polyamines/chemical synthesis , Polyamines/metabolism , Polyamines/pharmacology , Polyelectrolytes , Structure-Activity Relationship , beta-Cyclodextrins/chemical synthesis , beta-Cyclodextrins/metabolism , beta-Cyclodextrins/pharmacology
11.
Biophys J ; 99(3): 782-9, 2010 Aug 04.
Article in English | MEDLINE | ID: mdl-20682255

ABSTRACT

Using poly-(ethylene glycol)s of different molecular weights, we probe the channels formed in planar lipid bilayers by epsilon toxin secreted by the anaerobic bacterium Clostridium perfringens. We find that the pore is highly asymmetric. The cutoff size of polymers entering the pore through its opening from the cis side, the side of toxin addition, is approximately 500 Da, whereas the cutoff size for the polymers entering from the trans side is approximately 2300 Da. Comparing these characteristic molecular weights with those reported earlier for OmpF porin and the alpha-Hemolysin channel, we estimate the radii of cis and trans openings as 0.4 nm and 1.0 nm, respectively. The simplest geometry corresponding to these findings is that of a truncated cone. The asymmetry of the pore is also confirmed by measurements of the reversal potential at oppositely directed salt gradients. The moderate anionic selectivity of the channel is salted-out more efficiently when the salt concentration is higher at the trans side of the pore.


Subject(s)
Bacterial Toxins/pharmacology , Membranes, Artificial , Polyethylene Glycols/pharmacology , Action Potentials , Ion Channel Gating/physiology , Ions , Molecular Weight , Porosity/drug effects
12.
Biophys J ; 99(1): 134-43, 2010 Jul 07.
Article in English | MEDLINE | ID: mdl-20655841

ABSTRACT

Single channels of Bacillus anthracis protective antigen, PA(63), were reconstituted into planar lipid membranes and their inhibition by cationic aminopropylthio-beta-cyclodextrin, AmPrbetaCD, was studied. The design of the highly efficient inhibitor, the sevenfold symmetrical cyclodextrin molecule chemically modified to add seven positive charges, was guided by the symmetry and predominantly negative charge of the PA(63) pore. The protective action of this compound has been demonstrated earlier at both single-molecule and whole-organism levels. In this study, using noise analysis, statistics of time-resolved single-channel closure events, and multichannel measurements, we find that AmPrbetaCD action is bimodal. The inhibitor, when added to the cis side of the membrane, blocks the channel reversibly. At high salt concentrations, the AmPrbetaCD blockage of the channel is well described as a two-state Markov process, in which both the on- and off-rates are functions of the salt concentration, whereas the applied voltage affects only the off-rate. At salt concentrations smaller than 1.5 M, the second mode of AmPrbetaCD action on the channel is discovered: addition of the inhibitor enhances voltage gating, making the closed states of the channel more favorable. The effect depends on the lipid composition of the membrane.


Subject(s)
Antigens, Bacterial/metabolism , Bacterial Toxins/antagonists & inhibitors , Bacterial Toxins/metabolism , beta-Cyclodextrins/metabolism , beta-Cyclodextrins/pharmacology , Antigens, Bacterial/chemistry , Bacterial Toxins/chemistry , Cell Membrane/chemistry , Cell Membrane/drug effects , Cell Membrane/metabolism , Dose-Response Relationship, Drug , Electric Conductivity , Ion Channel Gating/drug effects , Kinetics , Lipid Metabolism , Markov Chains , Models, Molecular , Porosity , Protein Binding , Protein Conformation
13.
Antimicrob Agents Chemother ; 54(1): 298-304, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19805564

ABSTRACT

Staphylococcus aureus pneumonia is a common, potentially life-threatening infection caused by this human pathogen. The only therapies available to treat S. aureus pneumonia are antibiotics, a modality that is jeopardized by the organism's remarkable ability to acquire antimicrobial resistance. S. aureus alpha-hemolysin is a pore-forming cytotoxin that is essential for the pathogenesis of pneumonia. Strains lacking this cytotoxin are avirulent in a murine model of pneumonia; likewise, vaccine-based strategies that antagonize the toxin afford protection against lethal disease. Disruption of the function of this toxin therefore provides a potent mechanism to prevent and/or treat S. aureus pneumonia. beta-Cyclodextrin derivatives are small molecules with a sevenfold symmetry that mirrors the heptameric alpha-hemolysin. These compounds block the assembled alpha-hemolysin pore, compromising toxin function. We report that a modified beta-cyclodextrin compound, IB201, prevents alpha-hemolysin-induced lysis of human alveolar epithelial cells. This protective effect does not result from the ability of the beta-cyclodextrin to impair formation of the oligomeric alpha-hemolysin on the cell surface, supporting a role for this molecule in blockade of the lytic pore. An examination of IB201 in murine S. aureus pneumonia demonstrated that administration of this compound prevents and treats disease, protecting against mortality. Consistent with the vital importance of alpha-hemolysin in pneumonia caused by methicillin-sensitive and highly virulent methicillin-resistant S. aureus strains, IB201 protects against lethal challenge with both types of isolates. These observations, coupled with a favorable safety profile of beta-cyclodextrin compounds, provide a novel strategy that may be developed to combat S. aureus pneumonia.


Subject(s)
Pneumonia, Staphylococcal/drug therapy , beta-Cyclodextrins/therapeutic use , Animals , Cell Line , Cell Survival/drug effects , Erythrocytes/drug effects , Hemolysin Proteins , Hemolysis , Lung/microbiology , Methicillin-Resistant Staphylococcus aureus/drug effects , Mice , Mice, Inbred C57BL , Microbial Sensitivity Tests , Pneumonia, Staphylococcal/microbiology , Rabbits , Staphylococcus aureus/drug effects
14.
Antimicrob Agents Chemother ; 52(6): 2239-41, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18378717

ABSTRACT

We evaluated the in vivo efficacy of three beta-cyclodextrin derivatives that block the anthrax protective antigen pore. These compounds were at least 15-fold more potent than previously described beta-cyclodextrins in protecting against anthrax lethal toxin in a rat model. One of the drugs was shown to protect mice from bacterial infection.


Subject(s)
Anthrax/drug therapy , Anthrax/prevention & control , Bacillus anthracis/pathogenicity , Bacterial Toxins/antagonists & inhibitors , beta-Cyclodextrins/chemistry , beta-Cyclodextrins/pharmacology , Animals , Anthrax/mortality , Antigens, Bacterial/metabolism , Bacillus anthracis/drug effects , Bacterial Toxins/metabolism , Disease Models, Animal , Mice , Treatment Outcome
15.
Bioorg Med Chem ; 15(16): 5424-31, 2007 Aug 15.
Article in English | MEDLINE | ID: mdl-17572091

ABSTRACT

Many pathogens utilize the formation of transmembrane pores in target cells in the process of infection. A great number of pore-forming proteins, both bacterial and viral, are considered to be important virulence factors, which makes them attractive targets for the discovery of new therapeutic agents. Our research is based on the idea that compounds designed to block the pores can inhibit the action of virulence factors, and that the chances to find high affinity blocking agents increase if they have the same symmetry as the target pore. Recently, we demonstrated that derivatives of beta-cyclodextrin inhibited anthrax lethal toxin (LeTx) action by blocking the transmembrane pore formed by the protective antigen (PA) subunit of the toxin. To test the broader applicability of this approach, we sought beta-cyclodextrin derivatives capable of inhibiting the activity of Staphylococcus aureus alpha-hemolysin (alpha-HL), which is regarded as a major virulence factor playing an important role in staphylococcal infection. We identified several amino acid derivatives of beta-cyclodextrin that inhibited the activity of alpha-HL and LeTx in cell-based assays at low micromolar concentrations. One of the compounds was tested for the ability to block ion conductance through the pores formed by alpha-HL and PA in artificial lipid membranes. We anticipate that this approach can serve as the basis for a structure-directed drug discovery program to find new and effective therapeutics against various pathogens that utilize pore-forming proteins as virulence factors.


Subject(s)
Antigens, Bacterial/metabolism , Bacillus anthracis/metabolism , Bacterial Toxins/metabolism , Hemolysin Proteins/metabolism , Staphylococcus aureus/metabolism , beta-Cyclodextrins/chemistry , beta-Cyclodextrins/pharmacology , Animals , Electrophysiology , Erythrocytes/drug effects , Hemolysis/drug effects , Ions/chemistry , Mice , Models, Molecular , Molecular Structure , Rabbits , beta-Cyclodextrins/chemical synthesis
16.
Antimicrob Agents Chemother ; 51(1): 245-51, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17074791

ABSTRACT

In the course of Bacillus anthracis infection, B. anthracis lethal factor (LF) and edema factor bind to a protective antigen (PA) associated with cellular receptors ANTXR1 (TEM8) or ANTXR2 (CMG2), followed by internalization of the complex via receptor-mediated endocytosis. A new group of potential antianthrax drugs, beta-cyclodextrins, has recently been described. A member of this group, per-6-(3-aminopropylthio)-beta-cyclodextrin (AmPrbetaCD), was shown to inhibit the toxicity of LF in vitro and in vivo. In order to determine which steps in lethal factor trafficking are inhibited by AmPrbetaCD, we developed two targeted fluorescent tracers based on LFn, a catalytically inactive fragment of LF: (i) LFn site specifically labeled with the fluorescent dye AlexaFluor-594 (LFn-Al), and (ii) LFn-decorated liposomes loaded with the fluorescent dye 8-hydroxypyrene-1,3,6-trisulfonic acid (LFn-Lip). Both tracers retained high affinity to PA/ANTXR complexes and were readily internalized via receptor-mediated endocytosis. Using fluorescent microscopy, we found that AmPrbetaCD inhibits receptor-mediated cell uptake but not the binding of LFn-Al to PA/ANTXR complexes, suggesting that AmPrbetaCD works outside the cell. Moreover, AmPrbetaCD and LFn-Al synergistically protect RAW 264.7 cells from PA-mediated LF toxicity, confirming that AmPrbetaCD did not affect the binding of LFn-Al to receptor-associated PA. In contrast, AmPrbetaCD did not inhibit PA-mediated internalization of LFn-Lip, suggesting that multiplexing of LFn on the liposomal surface overcomes the inhibiting effects of AmPrbetaCD. Notably, internalized LFn-Al and LFn-Lip protected cells that overexpressed anthrax receptor TEM8 from PA-induced, LF-independent toxicity, suggesting an independent mechanism for PA inhibition inside the cell. These data suggest the potential for the use of beta-cyclodextrins in combination with LFn-Lip loaded with antianthrax drugs against intracellular targets.


Subject(s)
Antigens, Bacterial/metabolism , Bacillus anthracis/drug effects , Monocytes/drug effects , Peptide Fragments/pharmacology , Animals , Bacillus anthracis/growth & development , Bacillus anthracis/immunology , Bacterial Toxins/metabolism , CHO Cells , Cell Line , Cricetinae , Cricetulus , Dose-Response Relationship, Drug , Endocytosis/drug effects , Luminescent Proteins/chemical synthesis , Luminescent Proteins/chemistry , Luminescent Proteins/metabolism , Microscopy, Fluorescence , Monocytes/cytology , Monocytes/microbiology , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Protein Binding/drug effects , beta-Cyclodextrins/chemistry , beta-Cyclodextrins/metabolism , beta-Cyclodextrins/pharmacology
17.
Antimicrob Agents Chemother ; 50(11): 3740-53, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16982795

ABSTRACT

Recently, using structure-inspired drug design, we demonstrated that aminoalkyl derivatives of beta-cyclodextrin inhibited anthrax lethal toxin action by blocking the transmembrane pore formed by the protective antigen (PA) subunit of the toxin. In the present study, we evaluate a series of new beta-cyclodextrin derivatives with the goal of identifying potent inhibitors of anthrax toxins. Newly synthesized hepta-6-thioaminoalkyl and hepta-6-thioguanidinoalkyl derivatives of beta-cyclodextrin with alkyl spacers of various lengths were tested for the ability to inhibit cytotoxicity of lethal toxin in cells as well as to block ion conductance through PA channels reconstituted in planar bilayer lipid membranes. Most of the tested derivatives were protective against anthrax lethal toxin action at low or submicromolar concentrations. They also blocked ion conductance through PA channels at concentrations as low as 0.1 nM. The activities of the derivatives in both cell protection and channel blocking were found to depend on the length and chemical nature of the substituent groups. One of the compounds was also shown to block the edema toxin activity. It is hoped that these results will help to identify a new class of drugs for anthrax treatment, i.e., drugs that block the pathway for toxin translocation into the cytosol, the PA channel.


Subject(s)
Bacterial Toxins/antagonists & inhibitors , Cyclodextrins/chemical synthesis , Cyclodextrins/pharmacology , Animals , Anthrax/immunology , Antigens, Bacterial/immunology , Bacillus anthracis/immunology , CHO Cells , Cell Line , Cricetinae , Cyclic AMP/metabolism , Cyclodextrins/chemistry , Indicators and Reagents , Macrophages/drug effects , Macrophages/immunology , Magnetic Resonance Spectroscopy , Mice , Models, Molecular , Molecular Conformation , Neutralization Tests
18.
Bioorg Med Chem ; 14(1): 33-40, 2006 Jan 01.
Article in English | MEDLINE | ID: mdl-16169738

ABSTRACT

Recently, we demonstrated that simultaneous blocking of bacterial growth by antibiotics and inhibition of anthrax toxin action with antibodies against protective antigen were beneficial for the treatment of anthrax. The present study examined the hypothesis that blocking the pore formed by protective antigen can inhibit the action of anthrax toxin. The potential inhibitors were chosen by a structure-based design using beta-cyclodextrin as the starting molecule. Several beta-cyclodextrin derivatives were evaluated for their ability to protect RAW 264.7 cells from the action of anthrax lethal toxin. Per-substituted aminoalkyl derivatives displayed inhibitory activity and were protective against anthrax lethal toxin action at low micromolar concentrations. These results provide the basis for a structure-based drug discovery program, with the goal of identifying new drug candidates for anthrax treatment.


Subject(s)
Bacterial Toxins/antagonists & inhibitors , beta-Cyclodextrins/pharmacology , Animals , Antigens, Bacterial , Cell Line , Mice , beta-Cyclodextrins/chemistry
19.
Proc Natl Acad Sci U S A ; 102(42): 15075-80, 2005 Oct 18.
Article in English | MEDLINE | ID: mdl-16214885

ABSTRACT

Bacillus anthracis secretes three polypeptides: protective antigen (PA), lethal factor (LF), and edema factor (EF), which interact at the surface of mammalian cells to form toxic complexes. LF and EF are enzymes that target substrates within the cytosol; PA provides a heptameric pore to facilitate LF and EF transport into the cytosol. Other than administration of antibiotics shortly after exposure, there is currently no approved effective treatment for inhalational anthrax. Here we demonstrate an approach to disabling the toxin: high-affinity blockage of the PA pore by a rationally designed low-molecular weight compound that prevents LF and EF entry into cells. Guided by the sevenfold symmetry and predominantly negative charge of the PA pore, we synthesized small cyclic molecules of sevenfold symmetry, beta-cyclodextrins chemically modified to add seven positive charges. By channel reconstitution and high-resolution conductance recording, we show that per-6-(3-aminopropylthio)-beta-cyclodextrin interacts strongly with the PA pore lumen, blocking PA-induced transport at subnanomolar concentrations (in 0.1 M KCl). The compound protected RAW 264.7 mouse macrophages from cytotoxicity of anthrax lethal toxin (= PA + LF). More importantly, it completely protected the highly susceptible Fischer F344 rats from lethal toxin. We anticipate that this approach will serve as the basis for a structure-directed drug discovery program to find new and effective treatments for anthrax.


Subject(s)
Antigens, Bacterial/chemistry , Antigens, Bacterial/metabolism , Bacillus anthracis/metabolism , Bacterial Toxins/chemistry , Bacterial Toxins/metabolism , Drug Design , beta-Cyclodextrins/chemistry , beta-Cyclodextrins/metabolism , Animals , Anthrax/drug therapy , Antigens, Bacterial/genetics , Bacterial Toxins/genetics , Cell Line , Dose-Response Relationship, Drug , Electrophysiology , Humans , Macrophages/cytology , Macrophages/metabolism , Macrophages/microbiology , Male , Mice , Microbial Sensitivity Tests , Models, Molecular , Molecular Structure , Protein Conformation , Rats , Rats, Inbred F344 , beta-Cyclodextrins/therapeutic use
20.
FEMS Immunol Med Microbiol ; 40(1): 71-4, 2004 Jan 15.
Article in English | MEDLINE | ID: mdl-14734189

ABSTRACT

Currently there is no effective treatment for inhalational anthrax beyond administration of antibiotics shortly after exposure. There is need for new, safe and effective treatments to supplement traditional antibiotic therapy. Our study was based on the premise that simultaneous inhibition of lethal toxin action with antibodies and blocking of bacterial growth by antibiotics will be beneficial for the treatment of anthrax. In this study, we tested the effects of a combination treatment using purified rabbit or sheep anti-protective antigen (PA) antibodies and the antibiotic ciprofloxacin in a rodent anthrax model. In mice infected with a dose of Bacillus anthracis Sterne strain corresponding to 10 LD(50), antibiotic treatment with ciprofloxacin alone only cured 50% of infected animals. Administration of anti-PA IgG in combination with ciprofloxacin produced 90-100% survival. These data indicate that a combination of antibiotic/immunoglobulin therapy is more effective than antibiotic treatment alone in a rodent anthrax model.


Subject(s)
Anthrax/therapy , Antibodies, Bacterial/therapeutic use , Antigens, Bacterial , Bacterial Toxins/immunology , Ciprofloxacin/therapeutic use , Immunization, Passive , Animals , Anthrax/drug therapy , Anti-Infective Agents/therapeutic use , Antibodies, Bacterial/administration & dosage , Bacillus anthracis/drug effects , Bacillus anthracis/immunology , Enzyme-Linked Immunosorbent Assay , Female , Mice , Rabbits , Recombinant Proteins/immunology , Sheep
SELECTION OF CITATIONS
SEARCH DETAIL
...